Next Article in Journal
A Comparison of RML Prion Inactivation Efficiency by Heterogeneous and Homogeneous Photocatalysis
Previous Article in Journal
Dormancy-like Phenotype of Aggregatibacter actinomycetemcomitans: Survival during Famine
Previous Article in Special Issue
Periodontopathogens Porphyromonas gingivalis and Fusobacterium nucleatum and Their Roles in the Progression of Respiratory Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effect of Probiotic Supplements on the Oral Microbiota—A Narrative Review

by
Christine Lundtorp-Olsen
,
Merete Markvart
,
Svante Twetman
and
Daniel Belstrøm
*
Department of Odontology, Section for Clinical Oral Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
*
Author to whom correspondence should be addressed.
Pathogens 2024, 13(5), 419; https://doi.org/10.3390/pathogens13050419
Submission received: 23 April 2024 / Revised: 14 May 2024 / Accepted: 15 May 2024 / Published: 16 May 2024
(This article belongs to the Special Issue Insights in Oral Microbiota)

Abstract

:
Data from systematic reviews and meta-analyses show that probiotics positively impact clinical parameters of oral diseases such as gingivitis, dental caries, and periodontitis. However, the working mechanism of probiotics is not fully understood, but is hypothesized to be mediated by direct and indirect interactions with the oral microbiota and the human host. In the present narrative review, we focused on the microbiological effect of probiotic supplements based on data retrieved from randomized clinical trials (RCTs). In addition, we assessed to what extent contemporary molecular methods have been employed in clinical trials in the field of oral probiotics. Multiple RCTs have been performed studying the potential effect of probiotics on gingivitis, dental caries, and periodontitis, as evaluated by microbial endpoints. In general, results are conflicting, with some studies reporting a positive effect, whereas others are not able to record any effect. Major differences in terms of study designs and sample size, as well as delivery route, frequency, and duration of probiotic consumption, hamper comparison across studies. In addition, most RCTs have been performed with a limited sample size using relatively simple methods for microbial identification, such as culturing, qPCR, and DNA–DNA checkerboard, while high-throughput methods such as 16S sequencing have only been employed in a few studies. Currently, state-of-the-art molecular methods such as metagenomics, metatranscriptomics, and metaproteomics have not yet been used in RCTs in the field of probiotics. The present narrative review revealed that the effect of probiotic supplements on the oral microbiota remains largely uncovered. One important reason is that most RCTs are performed without studying the microbiological effect. To facilitate future systematic reviews and meta-analyses, an internationally agreed core outcome set for the reporting of microbial endpoints in clinical trials would be desirable. Such a standardized collection of outcomes would most likely improve the quality of probiotic research in the oral context.

1. Background

Oral health is shaped by the symbiotic relationship between the oral microbiota and the host [1], with the oral microbiota being critically involved in transitions from oral health towards the three major oral diseases—gingivitis, periodontitis, and dental caries [2,3,4]. Hence, treatment and prevention of oral diseases have traditionally had a strong microbiological focus, targeting oral biofilms [5,6].
Probiotics are defined by the World Health Organization (WHO) as live microorganisms which, when administrated in adequate amounts, confer health benefits to the host [7]. Probiotics, mainly Lactobacillus and Bifidobacterium species, have been demonstrated to have potential beneficial effects in the gastrointestinal area, including food allergy [8]. While the mode of action of probiotics is not completely understood in detail, one of the main desired actions is antimicrobial activity against pathogenic bacteria [9]. Hence, when considering the global burden of antibiotic resistance [10], probiotics are an attractive antimicrobial alternative to antibiotics.
Probiotics have been extensively tested in prevention and/or treatment of oral diseases, with results being conflicting. For example, two recent meta-analyses from 2022 and 2023 concluded that probiotics influenced clinical periodontal parameters such as bleeding on probing and probing pocket depth [11,12]. On the contrary, two other meta-analyses of ten and four clinical trials, respectively, did not show any effect of probiotics on bleeding on probing in patients with gingivitis [13,14]. In dental caries, a meta-analysis from 2023, based on 17 clinical trials, concluded that probiotics reduced incidence and progression of dental caries in preschool children [15], which is in concert with another meta-analysis from 2022 [16]. The major discrepancies observed in clinical parameters underscore the necessity to include microbiological endpoints in clinical studies, as knowledge on the microbiological mode of action of probiotics is essential for interpretation of the potential clinical effect observed.
In recent decades, there has been a tremendous development in molecular microbiological methods, which can be used to characterize the oral microbiota [17]. For example, moving from culturing procedures to culture-independent techniques, such as cloning and sequencing some 20 years ago, enabled identification of a substantial part of the uncultured and, therefore hitherto, unknown members of the oral microbiota [18]. Moreover, advancement to high-throughput next-generation sequencing methods facilitated taxonomic characterization of the oral microbiota in health and disease [19]. In continuation, metagenomic sequencing has enabled strain-level taxonomic resolution [20]. Also, advanced methods such as metatranscriptomics and metaproteomics have accelerated the transition from studies on microbial composition toward functional characterization of the oral microbiota [21,22]. Finally, whole-genome sequencing together with metagenome-assembled genomes provide the opportunity for detailed characterization of complete bacterial genomes [23,24]. As such, the molecular toolbox is now heavily equipped with the instruments needed for an in-depth analysis of probiotic strains and their potential impact on the oral microbiota. The question is whether these methods have found their way into the area of probiotics.
The purpose of the present study was to review the literature, with primary emphasis on the microbiological effect of probiotic supplements observed as compared to placebo in randomized clinical trials (RCTs). Secondly, the aim was to evaluate to which extent contemporary molecular methods are employed in clinical trials in the field of oral probiotics.

2. Periodontitis

Periodontitis is a multifactorial disease, which is the consequence of an imbalance of the oral microbiota and the host immune system, mediated by compositional changes of the subgingival microbiota and dysregulation of the host response, conditioned by heredity and environmental risk factors such as smoking. The ultimate oral consequence of periodontitis is tooth loss and edentulism, whereas periodontitis is also associated with increased risk of chronic inflammatory conditions such as type 2 diabetes and cardiovascular diseases [3]. Specifically, the subgingival environment in the established periodontal lesion is characterized by microbial dysbiosis, including compositional changes such as a higher abundance of proposed periodontal pathogens, which are predominantly Gram-negative anaerobic rods [25]. Along this line, studies have demonstrated that non-surgical periodontal treatments induce compositional changes to the subgingival microbiota, as illustrated by a higher abundance of Gram-positive rods and cocci in combination with a decrease in abundance of Gram-negative anaerobic bacteria [26,27]. As the most feasible application of probiotics in periodontal treatment is as a supplement to non-surgical treatment, probiotics should ideally augment the effect of this treatment on the subgingival microbiota.
In the last few decades, multiple RCTs with microbiological endpoints have been performed in periodontology ([28,29,30,31,32,33,34,35,36], Table 1), in which probiotic supplements were used either as an adjunct to non-surgical periodontal treatment [28,29,30,32,33,34,35,36], or without additional instrumentation [31]. In most cases, Lactobacillus and Bifidobacterium species were used as probiotic strains, being delivered in different forms, including mouthwash [28,36], capsules [29], suspensions [31], gels [30], and lozenges [30,32,34,35]. In addition to mode of delivery, there are other significant discrepancies in terms of sample size and study design, with some studies testing the short-term effect after 14 days [28] and others the long-term effect after 12 months [34]. Moreover, different microbiological methods have been used, including culturing, qPCR, and DNA–DNA checkerboard [28,29,30,31,32,33,34,35,36]. Naturally, the heterogenicity observed hampers the possibility of comparing data across the studies included.
From a microbiological point of view, the results seem conflicting, with some studies reporting no effect of the tested probiotic, irrespective of the microbiological endpoint [29,30,31,33,36]. Among the positive effects being reported, these include a significant decrease in Treponema denticola and Tannerella forsythia in the subgingival plaque as evaluated by PCR [28], a significant decrease in red complex bacteria in the subgingival plaque monitored by DNA–DNA checkerboard [32], a significant decrease in the percentage of obligate anaerobic bacteria in the subgingival plaque identified by culturing [34], and a significant reduction in salivary, supragingival and subgingival levels of Porphyromonas gingivalis as quantified by qPCR [35]. While the positive microbiological results can be seen as proof of principle, demonstrating an effect of the tested probiotic in vivo, it is important to remember that periodontitis is a complex disease with a polymicrobial etiology [25]. Hence, a probiotic impact evaluated solely as the effect on one or a few preselected proposed pathogens might not necessarily be of clinical importance.
From a technical perspective, the microbial methods employed for studies on probiotics in RCTs in periodontology are all relatively simple using either culturing techniques or early molecular methods, such as qPCR and DNA–DNA checkerboard. The common denominator of the methods used is that they are all close-ended, meaning that they are targeting a few specific proposed pathogens (culturing and qPCR) and up to a total of 40 pre-selected oral bacterial species (DNA–DNA checkerboard). To the best of our knowledge, no single study examining probiotics in periodontology has used high-throughput open-ended methods, such as 16S sequencing or metagenomics, which would have provided an in-depth characterization of the potential effect of probiotics on the subgingival microbiota, as expressed by alpha and beta diversity, as well as compositional changes. In addition, contemporary sophisticated methods, including metatranscriptomics and metaproteomics, have not been employed, which means that the impact of probiotics on the phenotypic profile of the subgingival microbiota in terms of functional information, such as gene expression, remains unknown. In the last decade, advanced molecular methods have found their way into studies on the general microbiology of periodontitis [19,37], providing detailed insight into the etiological role of the subgingival microbiota in health and disease. Importantly, the current perception of the role of the subgingival microbiota in the pathogenesis of periodontitis has greatly moved from a narrow focus on specific bacterial species toward a more comprehensive view on the total biofilm community, including synergistic and antagonistic interactions between members of the biofilm and interactions with the human host in different ecological conditions [25]. Preferably, future probiotic studies in periodontology with microbial endpoints should employ state-of-the-art molecular methods, providing detailed compositional and functional effects mediated by the probiotic tested.

3. Dental Caries

Dental caries is a complex disease, which in essence is the biochemical consequence of prolonged microbial carbohydrate metabolism, resulting in continuous pH drops in mature dental biofilms, facilitated by frequent exposure to dietary sugars [4,38]. Historically, the prime microbial focus in the field of dental caries has been on specific oral bacterial species with proficient carbohydrate metabolism, such as oral streptococci [39] and Lactobacillus species [40], with special emphasis on Streptococcus mutans due to the versatile armamentarium of caries-associated virulence factors [41]. In addition, studies have reported a positive correlation of salivary levels of S. mutans with caries experience [42], and salivary carriage of S. mutans has been suggested as a risk factor of future caries activity [43]. From a clinical perspective, probiotics could have multiple areas of application in the field of dental caries, but most importantly it would be suitable for non-invasive treatment of non-cavitated lesions, as well as in the prevention of the development of new lesions.
In the field of dental caries, a substantial number of RCTs have been performed testing the microbiological effect of probiotics in both children and adult populations ([44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61], Table 1). As is the case in periodontology, the probiotic strains used in cariology are almost exclusively Lactobacillus and Bifidobacterium species, being delivered in various ways, including mouthwash [44], yoghurt [45,46], milk [50,51,52,53,58,60,61], tablets [47,48,56], ice cream [49,59], oil [54], and cereals [55]. There are considerable differences in the RCTs with regard to study designs, with some studies evaluating the short-term effect after 7–14 days of consumption [44,45,47,51,60], and others the impact of long-term consumption between 6 and 9 months [50,52,61]. In addition, two studies have monitored the effect of consumption of probiotics during the first year after birth, in 9-year-olds [54,55]. In a substantial amount of these studies, culturing of S. mutans and/or Lactobacillus species from supragingival plaque and/or saliva samples was the only microbial analysis performed. Likewise, several studies have used chairside detection of S. mutans and/or Lactobacillus species. While very similar microbial endpoints provide the option for comparison across studies, these will obviously be heavily influenced by differences in study design, delivery modes and composition of cohorts.
In adults, a positive effect of probiotics, as evaluated by a significant decrease in S. mutans and/or Lactobacillus species in supragingival plaque and/or saliva samples, has been reported in multiple studies [19,44,45,46,47]. Importantly, studies reporting a positive microbiological effect in adults tested the short-term effect of the probiotics, as these were used for 14 days, with the microbial effect being evaluated after 14–30 days. In children and adolescents, the results are more diverging, with some studies reporting a positive effect of probiotic consumption on supragingival and salivary levels of S. mutans and/or Lactobacillus species [44,50,51,52,53,58,59,61], with other studies reporting no effect of the tested probiotic [54,55,56,57,60]. Notably, a common feature in studies reporting a positive effect was the evaluation of the probiotic effect immediately after short-term (7–14 days) [51,58,59], intermediate (3 months) [53] and long-term (6–9 months) [50,52] consumption of probiotics. In contrast, most studies showing no effect performed microbial evaluation several months to years after having stopped consuming the probiotic compound [54,55,56,57].
Collectively, studies which evaluated the microbial effect of probiotics immediately after a short, intermediate, or long-term consumption in children and adults were able to demonstrate an impact on supragingival and salivary levels of S. mutans and/or Lactobacillus species, which suggests that probiotic strains, including Lactobacillus and Bifidobacterium species, have a potential short-term impact on oral levels of proposed caries pathogens in the period of consumption. On the other hand, a persisting effect as evaluated months to years after consumption could not be detected. Hence, microbial data point towards the fact that prolonged consumption is needed to sustain a microbial effect of probiotics in the context of dental caries.
From a technical perspective, it is conspicuous that studies on the microbial effect of probiotics in the context of dental caries are based almost solely on culturing methods targeting proposed caries pathogens such as S. mutans and Lactobacillus species. Importantly, epidemiological studies have reported that while colonization with S. mutans is associated with increased risk of dental caries, S. mutans is not detected in a substantial part of dental caries cavities [62,63,64]. In addition, recent studies using contemporary molecular methods have demonstrated taxonomic and functional differences between the supragingival and salivary microbiota in dental caries versus oral health, which is not limited to S. mutans and Lactobacillus species [65,66,67]. Along this line, studies have demonstrated that other members of the oral microbiota, such as Veillonella species and Streptococcus sobrinus, may be better predictors of dental caries than S. mutans and Lactobacillus species [68,69]. Consequently, future probiotic studies in the field of dental caries that are performed using contemporary molecular methods are urgently needed, which will enable a shift in analysis towards focusing on taxonomic and functional characterization of the oral microbiota instead of the hitherto narrow focus on S. mutans and Lactobacillus species.

4. Gingivitis

Gingivitis is the most prevalent oral disease [70], with the microbial component as the central act in the pathogenesis of gingivitis being known since the 1960s [71]. Gingivitis, which is the consequence of undisturbed supragingival biofilm formation and maturation, is considered the predecessor of periodontitis [3], but not all cases of gingivitis will progress to periodontitis [72]. Due to its strong microbial etiology, prevention and treatment of gingivitis, i.e., professional dental cleaning, focus on supra- and subgingival plaque control. Hence, probiotics could be used to augment the microbiological effect of professional dental cleaning in the treatment of gingivitis.
Few RCTs have tested the microbiological effect of probiotics in the treatment of gingivitis ([73,74,75,76,77], Table 1). In gingivitis, different Lactobacillus species, including L. rhamnosus, L. curvatus, L. plantarum, L. brevis, and L. reuteri, have been delivered as tablets [74,75,76,77] or lozenges [73], either during experimental gingivitis [73,76] or as treatment of established gingivitis [74,75,77]. The microbiological effect has been evaluated in supragingival plaque [73,76], saliva [74], subgingival plaque [75], and simultaneously in subgingival plaque and saliva samples [77], using 16S sequencing [73,74], qPCR [75,77], and DNA–DNA checkerboard [76] immediately after probiotic consumption for 28 days to 8 weeks. In general, the comparable study designs, the almost similar study cohorts, as well as less heterogenicity in terms of delivery mode and duration of probiotic intake, assisted comparison of data across studies, while the use of different molecular methods together with different microbial samples being analyzed hampered comparison of data.
Microbiologically, some studies have reported the positive effects of probiotics on the microbial endpoints tested, including microbial resilience to experimental gingivitis in supragingival plaque [73], a significant reduction in subgingival levels of T. forsythia [75], and a significant reduction in P. gingivalis in subgingival plaque together with a significant reduction in total anaerobic counts and Prevotella intermedia in saliva [77]. In one study, no effect was observed on the composition of the salivary microbiota as evaluated by 16S sequencing [74], whereas another study failed to identify any effect on the supragingival microbiota during experimental gingivitis based on DNA–DNA checkerboard analysis [76].
As compared to research on probiotics in periodontology and cariology, two studies have employed modern high-throughput molecular methods for characterization of the salivary and the supragingival microbiota in gingivitis [73,74]. Hence, more detailed knowledge is available on the effect of these probiotic strains in the context of gingivitis, as compared to what could have been retrieved by culturing or use of close-ended methods targeting a limited number of pre-selected species. In addition, the use of 16S provided the opportunity to characterize the effect of the probiotics as evaluated by microbial diversities and relative abundances. Yet, sophisticated methods, such as metatranscriptomics and metaproteomics, which enable focus on bacterial functions and metabolic activity, rather than taxonomic composition, have not been used. Interestingly, a recent study demonstrated that virulence-related genes were upregulated in the transition from oral health to gingivitis, and that these changes were mediated by individual expression by specific bacterial species, underscoring the complexity of biofilm adaptation to the ecological changes accompanying the transition from health to gingivitis [78]. Along this line, two recent studies have reported different clinical trajectories of experimental gingivitis, which is not explained by the magnitude of clinical biofilm formation [79,80]. Taking these findings together, it is important that future studies testing the impact of probiotics on experimental gingivitis stratify and analyze the effect of the probiotic strains in individuals with different response patterns to experimental gingivitis, and subsequently use advanced molecular methods to illuminate bacterial gene expression inflicted by the probiotic tested.

5. Oral Health

The oral microbiota is the second most complex found in the human organism [81], with studies showing that the oral microbiota expresses both short- and long-term compositional stability if the ecological balance of the oral cavity is not disturbed [82,83]. On the contrary, external perturbations such as inadequate oral hygiene [84], frequent sugar intake [85], and use of systemic antibiotics [86] rapidly induce compositional changes to the oral microbiota. Hence, from a preventive perspective, if probiotics are to be used by orally healthy individuals, the aim should be to support compositional stability and resilience of the oral microbiota, when faced with stressful conditions.
Several probiotic RCTs with a microbial endpoint have been performed in orally healthy individuals ([87,88,89,90,91,92,93,94], Table 1), testing Lactobacillus, Bifidobacterium, and Streptococcus strains delivered as either tablets [91,92,93,94], lozenges [87,88,90] or gel [89] after consumption for 4–12 weeks. Microbial evaluation was performed immediately in either supragingival plaque, saliva, or subgingival plaque by means of different molecular methods, including 16S sequencing [87,88,89], qPCR [90,91,94], Human Oral Microbe Identification Microarray (HOMIM) [92], and DNA–DNA checkerboard [93]. The comparable study designs and the similar study cohorts together with the immediate evaluation of microbial endpoints facilitated the comparison of data, with different microbial identification methods and the use of various probiotic strains being the main confounding factors.
Table 1. Probiotic randomized clinical trials in periodontitis, dental caries, gingivitis and oral health.
Table 1. Probiotic randomized clinical trials in periodontitis, dental caries, gingivitis and oral health.
Author, Year, ReferenceCountrySample SizeProbiotic StrainsDelivery ModeMicrobial Sampling and AnalysisAuthors Reported Results
Periodontitis
Tapashetti et al., 2022, [28]IndiaN = 20Lactobacillus acidophilus
Lactobacillus rhamnosus
Bifidobacterium longum
Saccharomyces boulardii
Mouthwash
2 times per day, 14 days
qPCR
subgingival plaque
Significant decrease in Treponema denticola and Tannerella forsythia
De Oliveira et al., 2022, [29]BrazilN = 483 Lactobacillus spp.
2 Bifidobacterium spp.
Capsule
1 capsule per day, 30 days
DNA–DNA checkerboard
Subgingival plaque
No significant changes
Pudgar et al., 2021, [30]SloveniaN = 40Lactobacillus brevis
Lactobacillus plantarum
Gel and lozenges
1 time per day, 3 months
Culturing
Subgingival plaque
No significant changes
NĘdzi-GÓra et al., 2020, [31]PolandN = 51Lactobacillus salivarius SGL03Suspension
1 time per day, 30 days
Culturing
Supragingival plaque
No significant changes
Invernici et al., 2018, [32]BrazilN = 41Bifidobacterium animalis subsp. lactis (B. lactis) HN019Lozenges
1 time per day, 30 days
DNA–DNA checkerboard
Subgingival plaque
Significant decrease in red complex bacteria
Morales et al., 2018, [33]Chile N = 47Lactobacillus rhamnosus SP1Sachet
1 time per day, 3 months
DNA–DNA checkerboard
Culturing
Subgingival plaque
No significant changes
Tekce et al., 2015, [34]TurkeyN = 40Lactobacillus reuteriLozenges
2 times per day, 3 weeks
Culturing
Subgingival plaque
Significant decrease in % of obligate anaerobes
Teughels et al., 2013, [35]BelgiumN = 30Lactobacillus reuteriLozenges
2 times per day, 12 weeks
qPCR
Saliva, supragingival and subgingival plaque
Significant decrease in Porphyromonas gingivalis in saliva, supragingival and subgingival plaque
Tsubaru et al., 2009, [36]JapanN = 54Bacillus subtilisMouthwash
2 times per day, 1 month
BANA test/hybridization
Supragingival plaque
No significant changes
Dental Caries in adults
Krupa et al., 2022, [44]IndiaN = 30Lactobacillus acidophilus-R 0052
Lactobacillus rhamnosus-R 0011
Bifidobacterium longum-R 00175
Bacillus coagulans-SNZ 1969
Saccharomyces boulardii
Mouthwash
2 times per day, 14 days
Culturing
Supragingival plaque
Significant decrease in Streptococcus mutans
Javid et al., 2020, [45]IranN = 66Bifidobacterium lactis Bb12Yoghurt
1 time per day, 14 days
Culturing
Saliva
Significant decrease in Streptococcus mutans and Lactobacillus spp.
Ghamesi et al., 2017, [46]IranN = 50Lactobacillus acidophilusYoghurt
1 time per day, 3 weeks
Culturing
Saliva
Significant decrease in Streptococcus mutans
Nishihara et al., 2014, [47]JapanN = 64Lactobacillus salivarius WB21
Lactobacillus salivarius TI 2711
Tablets
3 times per day, 14 days
Culturing
Saliva
Significant decrease in Streptococcus mutans
Chuang et al., 2011, [48]ChinaN = 78Lactobacillus paracasei GMNL-33Tablets
3 timers per day, 14 days
SM and LB strip
Saliva
No significant changes
Caglar et al., 2008, [49]TurkeyN = 24Bifidobacterium animalis subsp. lactis BB-12Ice cream
1 time per day, 10 days
SM and LB strip
Saliva
Significant decrease in Streptococcus mutans
Dental Caries in children/adolescents
Krupa et al., 2022, [44]IndiaN = 30Lactobacillus acidophilus-R 0052
Lactobacillus rhamnosus-R 0011
Bifidobacterium longum-R 00175
Bacillus coagulans-SNZ 1969
Saccharomyces boulardii
Mouthwash
2 times per day, 14 days
Culturing
Supragingival plaque
Significant decrease in Streptococcus mutans
Manmontri et al., 2020, [50]ThailandN = 487Lactobacillus paracaseiMilk
3 times per week, 6 months
qPCR
Culturing
Saliva, supragingival plaque
Significant decrease in Streptococcus mutans and Lactobacillus spp.
Patil et al., 2019, [51]IndiaN = 30Lactobacillus caseiMilk
1 time per day, 7 days
Culturing
Saliva
Significant decrease in Streptococcus mutans
Villavicencio et al., 2018, [52]ColombiaN = 363Lactobacillus rhamnosus
Bifidobacterim longum
Milk
5 days per week, 9 months
Culturing
Saliva
Significant decrease in Lactobacillus spp.
Pahumunto et al., 2018, [53]ThailandN = 124Lactobacillus paracasei SD1Milk
1 time per day, 3 months
Culturing
Saliva
Significant decrease in Streptococcus mutans
Stensson et al., 2014, [54]SwedenN = 113Lactobacillus reuteri strain ATCC 55370Oil
5 drops per day, 1 year
Culturing
Saliva, supragingival plaque
No significant changes
Hasslöf et al., 2013, [55]SwedenN = 179Lactobacillus paracasei F19Cereals
1 time per day, 9 months
Culturing
Saliva
No significant changes
Taipale et al., 2013, [56]FinlandN = 163Bifidobacterium animalis subsp. lactis BB-12Tablets
1 time per day, 2 years
Culturing
SM strip
Supragingival plaque
No significant changes
Burton et al., 2013, [57]New ZealandN = 100Streptococcus salivarius M18Lozenges
2 times per day, 3 months
Culturing
Saliva
No significant changes
Juneja et al., 2012, [58]IndiaN = 40Lactobacillus rhamnosus hct 70Milk
1 time per day, 3 weeks
Culturing
Saliva
Significant decrease in Streptococcus mutans
Singh et al., 2011, [59]IndiaN = 40Bifidobacterium lactis Bb12
Lactobacillus acidophilus La5
Ice cream
1 time per day, 10 days
SM and LB strip
Saliva
Significant decrease in Streptococcus mutans
Lexner et al., 2010, [60]DenmarkN = 18Lactobacillus rhamnosus LB21Milk
1 time per day, 14 days
DNA–DNA checkerboard
Culturing
Saliva
No significant changes
Näse et al., 2001, [61]FinlandN = 594Lactobacillus rhamnosus GGMilk
5 days per week, 7 months
Culturing
Saliva, supragingival plaque
Significant decrease in Streptococcus mutans
Gingivitis
Lundtorp Olsen et al., 2023, [73]DenmarkN = 80Lactobacillus rhamnosus PB01, DSM 14869
Lactobacillus curvatus EB10, DSM 3230
Lozenges
2 times per day, 28 days
16S sequencing
Supragingival plaque
Significant impact on resilience of the supragingival microbiota
Keller et al., 2018, [74]DenmarkN = 47Lactobacillus rhamnosus PB01, DSM 14869
Lactobacillus curvatus EB10, DSM 3230
Tablets
2 times per day, 28 days
16S sequencing
Saliva
No significant changes
Montero et al., 2017, [75]SpainN = 59Lactobacillus plantarum,
Lactobacillus brevis and
Pediococcus acidilactici
Tablets
2 times per day, 6 weeks
qPCR
Subgingival plaque
Significant decrease in Tannerella forsythia
Hallström et al., 2013, [76]SwedenN = 18Lactobacillus reuteri (ATCC55730 and ATCC PTA5289)Tablets
2 times per day, 3 weeks
DNA–DNA checkerboard
Supragingival plaque
No significant changes
Iniesta et at., 2012, [77]SpainN = 40Lactobacillus reuteriTablets
1 time per day, 8 weeks
qPCR
Culturing
Significant decrease in Porphyromonas gingivalis and Prevotella intermedia
Oral Health
Lundtorp Olsen et al., 2021, [87]DenmarkN = 110Lactobacillus rhamnosus PB01, DSM 14869
Lactobacillus curvatus EB10, DSM 3230
Lozenges
2 times per day, 12 weeks
16S sequencing
Supragingival plaque
No significant changes
Lundtorp Olsen et al., 2021, [88]DenmarkN = 80Lactobacillus rhamnosus PB01, DSM 14869
Lactobacillus curvatus EB10, DSM 3230
Lozenges
2 times per day, 28 days
16S sequencing
Saliva
Significant decrease in Streptococcus spp.
Ferrer et al., 2020, [89]SpainN = 59Streptococcus dentisani 7746Gel
1 time per day, 1 months
16S sequencing
Supragingival plaque
Significant change in microbiota composition
Alanzi et al., 2018, [90]KuwaitN = 108Lactobacillus rhamnosus GG (LGG)
Bifidobacterium lactis BB-12
Lozenges
2 times per day, 4 weeks
qPCR
Saliva, supragingival plaque
Significant decrease in Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis and Fosubacterium nucleatum
Tobia et al., 2018, [91]JapanN = 16Lactobacillus crispatus KT-11 strain (KT-11)Tablets
1 time per day, 4 weeks
qPCR
Saliva
Significant decrease in Porphyromonas gingivalis
Toiviainen et al., 2015, [92]FinlandN = 60Lactobacillus rhamnosus GG
Bifidobacterium animalis subsp. lactis BB-12
Tablets
1 time per day, 4 weeks
HOMIM
Culturing
Saliva
No significant changes
Sinkiewitz, et al., 2010, [93]SwedenN = 23Lactobacillus reuteri ATCC 55730 and ATCC PTA 5289Tablets
1 time per day, 12 weeks
DNA–DNA checkerboard
Culturing
Saliva
No significant changes
Mayanagi et al., 2009, [94]JapanN = 66Lactobacillus salivarius WB21Tablets
1 time per day, 4 weeks
qPCR
Supragingival plaque
Significant decrease in periopathogens
Microbiologically, a handful of studies have reported a positive effect of the probiotic tested, such as significant compositional changes in the supragingival and salivary microbiota as characterized by 16S sequencing [88,89], and significant reductions in the proposed periodontal pathogens in plaque and saliva quantified by qPCR [90,91,94]. On the contrary, other studies failed to show an effect on supragingival plaque and saliva composition and levels of selected species as evaluated by means of 16S sequencing [87], HOMIM and culturing [93] and qPCR [94].
From a biological perspective, it is noteworthy that a considerable number of clinical trials performed in orally healthy individuals have focused primarily on the supragingival and salivary levels of proposed periodontal pathogens, when considering that salivary and supragingival carriage of these specific species is reported as relatively low in healthy adults [95,96,97,98]. Hence, one could argue that specific pathogenic species are not the most appropriate target for probiotics used in orally healthy individuals. In continuation, in two studies, where high-throughput sequencing demonstrated a significant impact of probiotics on the salivary [88] and the supragingival microbiota [89], the compositional changes were primarily driven by alterations in relative abundance of Streptococcus species in saliva, and supragingival abundance of the proposed cariogenic pathogen, Scardovia wiggsiae. While high-throughput molecular methods have already been used to study the effect of probiotics on the healthy oral microbiome, the working mechanisms remain to be uncovered, as no studies have employed methods which enable functional characterization of the microbiota.

6. Discussion

The present review of the literature has identified significant microbiological shortcomings in the research area of probiotics, as most RCTs do not have a microbiological endpoint. Indeed, this is a concern when considering that, from a theoretical perspective, some of the main proposed working mechanisms of probiotics, irrespective of body site, are direct and indirect interactions with the resident microbiota ([99,100], Figure 1). Arguably, microbiological data are therefore essential when interpreting clinical endpoints in clinical probiotic trials.
From a legislative point of view, probiotics are categorized as food supplements, which means that the extensive battery of rules and regulations from the pharmaceutical area does not apply to the probiotics industry. In other words, it is possible to produce and sell probiotics without having provided data showing the safety and clinical efficacy of the product. To the best of our knowledge, only one study has been conducted aimed specifically at testing the clinical and microbiological safety of a probiotic compound [87]. Certainly, this is surprising, considering that the global probiotics market is estimated to reach USD 85.4 billion in 2027 (https://www.marketsandmarkets.com/, accessed on 14 March 2024), meaning that it should be financially possible to thoroughly test probiotic products before being released to the market. Importantly, the effects on the oral microbiota of other oral care products, such as toothpaste and dentifrices, have been analyzed by means of advanced molecular methods, despite the fact that these products, like probiotics, are also not categorized as medical compounds [101,102]. Naturally, it is a great advantage that solid evidence is available, assisting dental professionals when they advise their patients which oral health care products to use, including probiotics.
Technically, the first step of screening for probiotic strains is performed using in vitro laboratory analysis, focusing on the effect of the probiotics on specific predefined microbial pathogens [103,104], which might explain why most probiotic RCTs solely investigate oral levels of specific predefined pathogens. While an effect observed in the laboratory is a prerequisite for further analysis, it is important to acknowledge that data generated using culturing and other in vitro setups represent a simplified version, as compared to the in vivo condition, where the probiotic will be in competition with the resident microbiota and influenced by the host. Hence, it is pivotal to address the impact of the probiotic not only on the preselected pathogens, but also on the total microbial community, requiring more sophisticated methods than culturing, qPCR, and DNA–DNA checkerboard, which until now are the techniques predominantly used for studies on probiotics in RCTs.
From a molecular perspective, oral health may be composed of different microbial and metabolomic profiles [105]. In addition, the composition of the oral microbiota is highly site-specific [106], and influenced not only by oral health status, but also by general medical disorders and age [107,108,109]. Moreover, frequency and mode of delivery are of critical importance, as probiotic supplements will most likely have the most pronounced effect in situations where the oral biofilm is also being mechanically disrupted. In addition, the probiotic supplement should ideally be present in the oral cavity for a prolonged time to have maximal effect. Hence, the ideal frequency and mode of delivery will most likely not be the same in the context of dental caries, gingivitis, and periodontitis. Consequently, it is critically important when choosing a probiotic to counterweigh the expected beneficial effects at diseased sites against potential adverse effects at other oral sites, or in predisposed individuals. With that in mind, it is staggering that almost identical probiotic strains have been tested in both periodontitis and caries, when considering that proposed pathogens of the two diseases are critically different in terms of their ecological preferences (pH and O2) and metabolic profile [110]. Hence, from a theoretical point of view, caries probiotics could potentially favor periodontal pathogens and vice versa. Importantly, epidemiological evidence suggests different individual predispositions to the development of dental caries and periodontitis [111], which is in line with recent data on experimental gingivitis, showing different inflammatory reaction patterns to biofilm formation [79,80]. Hence, from a biological perspective, we speculate that the same probiotic strain could have different microbiological effects based on parameters such as baseline microbial composition, age, gender, as well as oral and general health status. Accordingly, this call for action, with various probiotics being used in individuals with different oral health risk profiles, is part of an individualized oral precision medicine strategy, as known from other areas such as oncology [112].
A substantial number of probiotic studies focus on the abundance of specific predefined pathogens, such as S. mutans and P. gingivalis, thereby adhering to key elements of the specific plaque hypothesis, which was rejected in the mid-1990s and substituted by the ecological plaque hypothesis [113]. As initiated by the red complex theory [114], and further developed by the keystone pathogen hypothesis [115], P. gingivalis has attracted considerable attention as an etiological agent of periodontitis, which is biologically grounded, as P. gingivalis possesses a wide variety of periodontitis-associated virulence factors [116]. While the former perception of P. gingivalis was that virulence was primarily the consequence of high subgingival abundance, recent literature using state-of-the-art molecular methods points towards P. gingivalis being highly pathogenic even in low numbers, as the pathogenicity is mediated through interactions with the resident microbiota and the human complement system [117,118]. Consequently, P. gingivalis can potentially still orchestrate prolonged disease activity, despite being deprived in number by a probiotic. However, this will not be identified using simple molecular methods focusing solely on levels of P. gingivalis or other specific bacteria. Interestingly, recent literature has employed metatranscriptomics to portray in detail microbial activity in periodontitis, as quantified not only by bacterial gene expression of P. gingivalis [119], but also resident members of the oral microbiota such as oral streptococci [37]. As such, contemporary data could be used in future development of next-generation probiotics in periodontology, focusing on both depressing pathogenic gene expression and augmenting natural counterbalancing gene expression of the resident oral microbiota.
The focus of the present review is solely on the effect of probiotic supplements on the oral microbiota, which is why studies on prebiotics, synbiotics, and postbiotics were not included. However, it is important to stress that studies have demonstrated the potential of using prebiotics such as arginine and non-cariogenic sugars in the prevention of dental caries [120,121], as well as dietary fibers in the prevention of periodontitis [122], thereby illuminating a preventive potential of prebiotics in oral care.

7. Concluding Remarks

One of the main expected working mechanisms of probiotics is through direct and indirect interactions with the resident oral microbiota. Yet, most clinical oral probiotic RCTs have not addressed the microbial effect of the probiotic tested. Hence, to facilitate future systematic reviews and meta-analyses, microbial endpoints should ideally be considered mandatory in all probiotic clinical trials. In addition, an internationally agreed best practice guideline on clinical trials on oral probiotics should be developed by the probiotic scientific community, inspired by important guidelines such as the STROBE guidelines [123] and the PRISMA guidelines [124]. Setting an international standard in terms of study design, with time of delivery as well as mode of delivery depending on the clinical condition, as well as core outcomes for the reporting of microbial endpoints in clinical trials, would be desirable, as such a standardized international guideline would most likely improve the quality of probiotic research in the oral context.

Author Contributions

Conceptualization, C.L.-O., M.M., S.T. and D.B.; formal analysis, C.L.-O. and D.B.; writing—original draft preparation, D.B.; writing—review and editing, M.M., S.T. and C.L.-O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Marsh, P.D.; Zaura, E. Dental biofilm: Ecological interactions in health and disease. J. Clin. Periodontol. 2017, 44 (Suppl. S18), S12–S22. [Google Scholar] [CrossRef] [PubMed]
  2. Abusleme, L.; Hoare, A.; Hong, B.Y.; Diaz, P.I. Microbial signatures of health, gingivitis, and periodontitis. Periodontol 2000 2021, 86, 57–78. [Google Scholar] [CrossRef]
  3. Van Dyke, T.E.; Bartold, P.M.; Reynolds, E.C. The Nexus between Periodontal Inflammation and Dysbiosis. Front. Immunol. 2020, 11, 511. [Google Scholar] [CrossRef] [PubMed]
  4. Pitts, N.B.; Zero, D.T.; Marsh, P.D.; Ekstrand, K.; Weintraub, J.A.; Ramos-Gomez, F.; Tagami, J.; Twetman, S.; Tsakos, G.; Ismail, A. Dental caries. Nat. Rev. Dis. Primers. 2017, 3, 17030. [Google Scholar] [CrossRef] [PubMed]
  5. Chapple, I.L.; Van der Weijden, F.; Doerfer, C.; Herrera, D.; Shapira, L.; Polak, D.; Madianos, P.; Louropoulou, A.; Machtei, E.; Donos, N.; et al. Primary prevention of periodontitis: Managing gingivitis. J. Clin. Periodontol. 2015, 42 (Suppl. S16), S71–S76. [Google Scholar] [CrossRef] [PubMed]
  6. Veiga, N.; Figueiredo, R.; Correia, P.; Lopes, P.; Couto, P.; Fernandes, G.V.O. Methods of Primary Clinical Prevention of Dental Caries in the Adult Patient: An Integrative Review. Healthcare 2023, 11, 1635. [Google Scholar] [CrossRef] [PubMed]
  7. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed]
  8. Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 605–616. [Google Scholar] [CrossRef] [PubMed]
  9. Kechagia, M.; Basoulis, D.; Konstantopoulou, S.; Dimitriadi, D.; Gyftopoulou, K.; Skarmoutsou, N.; Fakiri, E.M. Health benefits of probiotics: A review. ISRN Nutr. 2013, 2013, 481651. [Google Scholar] [CrossRef] [PubMed]
  10. Antimicrobial Resistance Collaborators. Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef] [PubMed]
  11. Gheisary, Z.; Mahmood, R.; Harri Shivanantham, A.; Liu, J.; Lieffers, J.R.L.; Papagerakis, P.; Papagerakis, S. The Clinical, Microbiological, and Immunological Effects of Probiotic Supplementation on Prevention and Treatment of Periodontal Diseases: A Systematic Review and Meta-Analysis. Nutrients 2022, 14, 1036. [Google Scholar] [CrossRef] [PubMed]
  12. Li, J.; Zhao, G.; Zhang, H.M.; Zhu, F.F. Probiotic adjuvant treatment in combination with scaling and root planing in chronic periodontitis: A systematic review and meta-analysis. Benef. Microbes 2023, 14, 95–107. [Google Scholar] [CrossRef]
  13. Akram, Z.; Shafqat, S.S.; Aati, S.; Kujan, O.; Fawzy, A. Clinical efficacy of probiotics in the treatment of gingivitis: A systematic review and meta-analysis. Aust. Dent. J. 2020, 65, 12–20. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, W.; Ren, J.; Li, J.; Peng, S.; Zhang, C.; Lin, Y. Effects of probiotics on the oral health of patients undergoing orthodontic treatment: A systematic review and meta-analysis. Eur. J. Orthod. 2023, 45, 599–611. [Google Scholar] [CrossRef] [PubMed]
  15. Meng, N.; Liu, Q.; Dong, Q.; Gu, J.; Yang, Y. Effects of probiotics on preventing caries in preschool children: A systematic review and meta-analysis. J. Clin. Pediatr. Dent. 2023, 47, 85–100. [Google Scholar] [CrossRef]
  16. Shi, J.; Wang, Q.; Ruan, G.; Chen, Y.; Zhao, M.; Shi, D.; Pan, B.; Xu, Z.; Zhang, T.; Wang, F.; et al. Efficacy of probiotics against dental caries in children: A systematic review and meta-analysis. Crit. Rev. Food Sci. Nutr. 2023, 63, 9977–9994. [Google Scholar] [CrossRef]
  17. Krishnan, K.; Chen, T.; Paster, B.J. A practical guide to the oral microbiome and its relation to health and disease. Oral Dis. 2017, 23, 276–286. [Google Scholar] [CrossRef]
  18. Paster, B.J.; Boches, S.K.; Galvin, J.L.; Ericson, R.E.; Lau, C.N.; Levanos, V.A.; Sahasrabudhe, A.; Dewhirst, F.E. Bacterial diversity in human subgingival plaque. J. Bacteriol. 2001, 183, 3770–3783. [Google Scholar] [CrossRef] [PubMed]
  19. Kumar, P.S.; Dabdoub, S.M.; Ganesan, S.M. Probing periodontal microbial dark matter using metataxonomics and metagenomics. Periodontology 2000 2021, 85, 12–27. [Google Scholar] [CrossRef] [PubMed]
  20. Valles-Colomer, M.; Blanco-Míguez, A.; Manghi, P.; Asnicar, F.; Dubois, L.; Golzato, D.; Armanini, F.; Cumbo, F.; Huang, K.D.; Manara, S.; et al. The person-to-person transmission landscape of the gut and oral microbiomes. Nature 2023, 614, 125–135. [Google Scholar] [CrossRef] [PubMed]
  21. Duran-Pinedo, A.E. Metatranscriptomic analyses of the oral microbiome. Periodontology 2000 2021, 85, 28–45. [Google Scholar] [CrossRef] [PubMed]
  22. Grassl, N.; Kulak, N.A.; Pichler, G.; Geyer, P.E.; Jung, J.; Schubert, S.; Sinitcyn, P.; Cox, J.; Mann, M. Ultra-deep and quantitative saliva proteome reveals dynamics of the oral microbiome. Genome Med. 2016, 8, 44. [Google Scholar] [CrossRef] [PubMed]
  23. Caselli, E.; Fabbri, C.; D’Accolti, M.; Soffritti, I.; Bassi, C.; Mazzacane, S.; Franchi, M. Defining the oral microbiome by whole-genome sequencing and resistome analysis: The complexity of the healthy picture. BMC Microbiol. 2020, 20, 120. [Google Scholar] [CrossRef] [PubMed]
  24. Setubal, J.C. Metagenome-assembled genomes: Concepts, analogies, and challenges. Biophys. Rev. 2021, 13, 905–909. [Google Scholar] [CrossRef] [PubMed]
  25. Belibasakis, G.N.; Belstrøm, D.; Eick, S.; Gursoy, U.K.; Johansson, A.; Könönen, E. Periodontal microbiology and microbial etiology of periodontal diseases: Historical concepts and contemporary perspectives. Periodontology 2000, 2023; Epub ahead of print. [Google Scholar] [CrossRef]
  26. Chen, C.; Hemme, C.; Beleno, J.; Shi, Z.J.; Ning, D.; Qin, Y.; Tu, Q.; Jorgensen, M.; He, Z.; Wu, L.; et al. Oral microbiota of periodontal health and disease and their changes after nonsurgical periodontal therapy. ISME J. 2018, 12, 1210–1224. [Google Scholar] [CrossRef] [PubMed]
  27. Byrne, S.J.; Chang, D.; Adams, G.G.; Butler, C.A.; Reynolds, E.C.; Darby, I.B.; Dashper, S.G. Microbiome profiles of non-responding and responding paired periodontitis sites within the same participants following non-surgical treatment. J. Oral Microbiol. 2022, 14, 2043595. [Google Scholar] [CrossRef] [PubMed]
  28. Tapashetti, R.P.; Ansari, M.W.; Fatima, G.; Bhutani, N.; Sameen, N.; Hm, P. Effects of Probiotics Mouthwash on Levels of Red Complex Bacteria in Chronic Periodontitis Patients: A Clinico-microbiological Study. J. Contemp. Dent. Pract. 2022, 23, 320–326. [Google Scholar] [CrossRef] [PubMed]
  29. De Oliveira, A.M.; Lourenço, T.G.B.; Colombo, A.P.V. Impact of systemic probiotics as adjuncts to subgingival instrumentation on the oral-gut microbiota associated with periodontitis: A randomized controlled clinical trial. J. Periodontol. 2022, 93, 31–44. [Google Scholar] [CrossRef] [PubMed]
  30. Pudgar, P.; Povšič, K.; Čuk, K.; Seme, K.; Petelin, M.; Gašperšič, R. Probiotic strains of Lactobacillus brevis and Lactobacillus plantarum as adjunct to non-surgical periodontal therapy: 3-month results of a randomized controlled clinical trial. Clin. Oral Investig. 2021, 25, 1411–1422. [Google Scholar] [CrossRef] [PubMed]
  31. NĘdzi-GÓra, M.; WrÓblewska, M.; GÓrska, R. The Effect of Lactobacillus salivarius SGL03 on Clinical and Microbiological Parameters in Periodontal Patients. Pol. J. Microbiol. 2020, 69, 441–451. [Google Scholar] [CrossRef] [PubMed]
  32. Invernici, M.M.; Salvador, S.L.; Silva, P.H.; Soares, M.S.; Casarin, R.; Palioto, D.B.; Souza, S.L.; Taba, M., Jr.; Novaes, A.B., Jr.; Furlaneto, F.A.; et al. Effects of Bifidobacterium probiotic on the treatment of chronic periodontitis: A randomized clinical trial. J. Clin. Periodontol. 2018, 45, 1198–1210. [Google Scholar] [CrossRef] [PubMed]
  33. Morales, A.; Gandolfo, A.; Bravo, J.; Carvajal, P.; Silva, N.; Godoy, C.; Garcia-Sesnich, J.; Hoare, A.; Diaz, P.; Gamonal, J. Microbiological and clinical effects of probiotics and antibiotics on nonsurgical treatment of chronic periodontitis: A randomized placebo-controlled trial with 9-month follow-up. J. Appl. Oral Sci. 2018, 26, e20170075. [Google Scholar] [CrossRef] [PubMed]
  34. Tekce, M.; Ince, G.; Gursoy, H.; Dirikan Ipci, S.; Cakar, G.; Kadir, T.; Yılmaz, S. Clinical and microbiological effects of probiotic lozenges in the treatment of chronic periodontitis: A 1-year follow-up study. J. Clin. Periodontol. 2015, 42, 363–372. [Google Scholar] [CrossRef] [PubMed]
  35. Teughels, W.; Durukan, A.; Ozcelik, O.; Pauwels, M.; Quirynen, M.; Haytac, M.C. Clinical and microbiological effects of Lactobacillus reuteri probiotics in the treatment of chronic periodontitis: A randomized placebo-controlled study. J. Clin. Periodontol. 2013, 40, 1025–1035. [Google Scholar] [CrossRef] [PubMed]
  36. Tsubura, S.; Mizunuma, H.; Ishikawa, S.; Oyake, I.; Okabayashi, M.; Katoh, K.; Shibata, M.; Iizuka, T.; Toda, T.; Iizuka, T. The effect of Bacillus subtilis mouth rinsing in patients with periodontitis. Eur. J. Clin. Microbiol. Infect. Dis. 2009, 28, 1353–1356. [Google Scholar] [CrossRef] [PubMed]
  37. Belstrøm, D.; Constancias, F.; Drautz-Moses, D.I.; Schuster, S.C.; Veleba, M.; Mahé, F.; Givskov, M. Periodontitis associates with species-specific gene expression of the oral microbiota. NPJ Biofilms Microbiomes 2021, 7, 76. [Google Scholar] [CrossRef]
  38. Selwitz, R.H.; Ismail, A.I.; Pitts, N.B. Dental caries. Lancet 2007, 369, 51–59. [Google Scholar] [CrossRef]
  39. Abranches, J.; Zeng, L.; Kajfasz, J.K.; Palmer, S.R.; Chakraborty, B.; Wen, Z.T.; Richards, V.P.; Brady, L.J.; Lemos, J.A. Biology of Oral Streptococci. Microbiol Spectr. 2018, 6. [Google Scholar] [CrossRef]
  40. Caufield, P.W.; Schön, C.N.; Saraithong, P.; Li, Y.; Argimón, S. Oral Lactobacilli and Dental Caries: A Model for Niche Adaptation in Humans. J. Dent. Res. 2015, 94 (Suppl. S9), 110S–118S. [Google Scholar] [CrossRef]
  41. Lemos, J.A.; Palmer, S.R.; Zeng, L.; Wen, Z.T.; Kajfasz, J.K.; Freires, I.A.; Abranches, J.; Brady, L.J. The Biology of Streptococcus mutans. Microbiol. Spectr. 2019, 7. [Google Scholar] [CrossRef]
  42. Pannu, P.; Gambhir, R.; Sujlana, A. Correlation between the salivary Streptococcus mutans levels and dental caries experience in adult population of Chandigarh, India. Eur. J. Dent. 2013, 7, 191–195. [Google Scholar] [CrossRef] [PubMed]
  43. Hemadi, A.S.; Huang, R.; Zhou, Y.; Zou, J. Salivary proteins and microbiota as biomarkers for early childhood caries risk assessment. Int. J. Oral Sci. 2017, 9, e1. [Google Scholar] [CrossRef]
  44. Krupa, N.C.; Thippeswamy, H.M.; Chandrashekar, B.R. Antimicrobial efficacy of Xylitol, Probiotic and Chlorhexidine mouth rinses among children and elderly population at high risk for dental caries—A Randomized Controlled Trial. J. Prev. Med. Hyg. 2022, 63, E282–E287. [Google Scholar] [CrossRef] [PubMed]
  45. Zare Javid, A.; Amerian, E.; Basir, L.; Ekrami, A.; Haghighizadeh, M.H.; Maghsoumi-Norouzabad, L. Effects of the Consumption of Probiotic Yogurt Containing Bifidobacterium lactis Bb12 on the Levels of Streptococcus mutans and Lactobacilli in Saliva of Students with Initial Stages of Dental Caries: A Double-Blind Randomized Controlled Trial. Caries Res. 2020, 54, 68–74. [Google Scholar] [CrossRef] [PubMed]
  46. Ghasemi, E.; Mazaheri, R.; Tahmourespour, A. Effect of Probiotic Yogurt and Xylitol-Containing Chewing Gums on Salivary S Mutans Count. J. Clin. Pediatr. Dent. 2017, 41, 257–263. [Google Scholar] [CrossRef] [PubMed]
  47. Nishihara, T.; Suzuki, N.; Yoneda, M.; Hirofuji, T. Effects of Lactobacillus salivarius-containing tablets on caries risk factors: A randomized open-label clinical trial. BMC Oral Health 2014, 14, 110. [Google Scholar] [CrossRef] [PubMed]
  48. Chuang, L.C.; Huang, C.S.; Ou-Yang, L.W.; Lin, S.Y. Probiotic Lactobacillus paracasei effect on cariogenic bacterial flora. Clin. Oral Investig. 2011, 15, 47. [Google Scholar] [CrossRef] [PubMed]
  49. Caglar, E.; Kuscu, O.O.; Selvi Kuvvetli, S.; Kavaloglu Cildir, S.; Sandalli, N.; Twetman, S. Short-term effect of ice-cream containing Bifidobacterium lactis Bb-12 on the number of salivary mutans streptococci and lactobacilli. Acta Odontol. Scand. 2008, 66, 154–158. [Google Scholar] [CrossRef] [PubMed]
  50. Manmontri, C.; Nirunsittirat, A.; Piwat, S.; Wattanarat, O.; Pahumunto, N.; Makeudom, A.; Sastraruji, T.; Krisanaprakornkit, S.; Teanpaisan, R. Reduction of Streptococcus mutans by probiotic milk: A multicenter randomized controlled trial. Clin. Oral Investig. 2020, 24, 2363–2374. [Google Scholar] [CrossRef]
  51. Patil, R.U.; Dastoor, P.P.; Unde, M.P. Comparative evaluation of antimicrobial effectiveness of probiotic milk and fluoride mouthrinse on salivary Streptococcus mutans counts and plaque scores in children—An in vivo experimental study. J. Indian Soc. Pedod. Prev. Dent. 2019, 37, 378–382. [Google Scholar] [CrossRef] [PubMed]
  52. Villavicencio, J.; Villegas, L.M.; Arango, M.C.; Arias, S.; Triana, F. Effects of a food enriched with probiotics on Streptococcus mutans and Lactobacillus spp. salivary counts in preschool children: A cluster randomized trial. J. Appl. Oral Sci. 2018, 26, e20170318. [Google Scholar] [CrossRef] [PubMed]
  53. Pahumunto, N.; Piwat, S.; Chankanka, O.; Akkarachaneeyakorn, N.; Rangsitsathian, K.; Teanpaisan, R. Reducing mutans streptococci and caries development by Lactobacillus paracasei SD1 in preschool children: A randomized placebo-controlled trial. Acta Odontol. Scand. 2018, 76, 331–337. [Google Scholar] [CrossRef]
  54. Stensson, M.; Koch, G.; Coric, S.; Abrahamsson, T.R.; Jenmalm, M.C.; Birkhed, D.; Wendt, L.K. Oral administration of Lactobacillus reuteri during the first year of life reduces caries prevalence in the primary dentition at 9 years of age. Caries Res. 2014, 48, 111–117. [Google Scholar] [CrossRef] [PubMed]
  55. Hasslöf, P.; West, C.E.; Videhult, F.K.; Brandelius, C.; Stecksén-Blicks, C. Early intervention with probiotic Lactobacillus paracasei F19 has no long-term effect on caries experience. Caries Res. 2013, 47, 559–565. [Google Scholar] [CrossRef] [PubMed]
  56. Taipale, T.; Pienihäkkinen, K.; Alanen, P.; Jokela, J.; Söderling, E. Administration of Bifidobacterium animalis subsp. lactis BB-12 in early childhood: A post-trial effect on caries occurrence at four years of age. Caries Res. 2013, 47, 364–372. [Google Scholar] [CrossRef] [PubMed]
  57. Burton, J.P.; Drummond, B.K.; Chilcott, C.N.; Tagg, J.R.; Thomson, W.M.; Hale, J.D.F.; Wescombe, P.A. Influence of the probiotic Streptococcus salivarius strain M18 on indices of dental health in children: A randomized double-blind, placebo-controlled trial. J. Med. Microbiol. 2013, 62 Pt 6, 875–884. [Google Scholar] [CrossRef] [PubMed]
  58. Juneja, A.; Kakade, A. Evaluating the effect of probiotic containing milk on salivary mutans streptococci levels. J. Clin. Pediatr. Dent. 2012, 37, 9–14. [Google Scholar] [CrossRef] [PubMed]
  59. Singh, R.P.; Damle, S.G.; Chawla, A. Salivary mutans streptococci and lactobacilli modulations in young children on consumption of probiotic ice-cream containing Bifidobacterium lactis Bb12 and Lactobacillus acidophilus La5. Acta Odontol. Scand. 2011, 69, 389–394. [Google Scholar] [CrossRef] [PubMed]
  60. Lexner, M.O.; Blomqvist, S.; Dahlén, G.; Twetman, S. Microbiological profiles in saliva and supragingival plaque from caries-active adolescents before and after a short-term daily intake of milk supplemented with probiotic bacteria—A pilot study. Oral Health Prev. Dent. 2010, 8, 383–388. [Google Scholar] [PubMed]
  61. Näse, L.; Hatakka, K.; Savilahti, E.; Saxelin, M.; Pönkä, A.; Poussa, T.; Korpela, R.; Meurman, J.H. Effect of long-term consumption of a probiotic bacterium, Lactobacillus rhamnosus GG, in milk on dental caries and caries risk in children. Caries Res. 2001, 35, 412–420. [Google Scholar] [CrossRef] [PubMed]
  62. Okada, M.; Kawamura, M.; Oda, Y.; Yasuda, R.; Kojima, T.; Kurihara, H. Caries prevalence associated with Streptococcus mutans and Streptococcus sobrinus in Japanese schoolchildren. Int. J. Paediatr. Dent. 2012, 22, 342–348. [Google Scholar] [CrossRef] [PubMed]
  63. Oda, Y.; Hayashi, F.; Okada, M. Longitudinal study of dental caries incidence associated with Streptococcus mutans and Streptococcus sobrinus in patients with intellectual disabilities. BMC Oral Health 2015, 15, 102. [Google Scholar] [CrossRef] [PubMed]
  64. Köhler, B.; Andréen, I. Mutans streptococci and caries prevalence in children after early maternal caries prevention: A follow-up at eleven and fifteen years of age. Caries Res. 2010, 44, 453–458. [Google Scholar] [CrossRef] [PubMed]
  65. Butcher, M.C.; Short, B.; Veena, C.L.R.; Bradshaw, D.; Pratten, J.R.; McLean, W.; Shaban, S.M.A.; Ramage, G.; Delaney, C. Meta-analysis of caries microbiome studies can improve upon disease prediction outcomes. APMIS 2022, 130, 763–777. [Google Scholar] [CrossRef] [PubMed]
  66. Hurley, E.; Barrett, M.P.J.; Kinirons, M.; Whelton, H.; Ryan, C.A.; Stanton, C.; Harris, H.M.B.; O’Toole, P.W. Comparison of the salivary and dentinal microbiome of children with severe-early childhood caries to the salivary microbiome of caries-free children. BMC Oral Health 2019, 19, 13. [Google Scholar] [CrossRef] [PubMed]
  67. Yang, F.; Zeng, X.; Ning, K.; Liu, K.L.; Lo, C.C.; Wang, W.; Chen, J.; Wang, D.; Huang, R.; Chang, X.; et al. Saliva microbiomes distinguish caries-active from healthy human populations. ISME J. 2012, 6, 1–10. [Google Scholar] [CrossRef] [PubMed]
  68. Gross, E.L.; Beall, C.J.; Kutsch, S.R.; Firestone, N.D.; Leys, E.J.; Griffen, A.L. Beyond Streptococcus mutans: Dental caries onset linked to multiple species by 16S rRNA community analysis. PLoS ONE 2012, 7, e47722. [Google Scholar] [CrossRef] [PubMed]
  69. Conrads, G.; de Soet, J.J.; Song, L.; Henne, K.; Sztajer, H.; Wagner-Döbler, I.; Zeng, A.P. Comparing the cariogenic species Streptococcus sobrinus and S. mutans on whole genome level. J. Oral Microbiol. 2014, 6, 26189. [Google Scholar] [CrossRef] [PubMed]
  70. Chapple, I.L.C.; Mealey, B.L.; Van Dyke, T.E.; Bartold, P.M.; Dommisch, H.; Eickholz, P.; Geisinger, M.L.; Genco, R.J.; Glogauer, M.; Goldstein, M.; et al. Periodontal health and gingival diseases and conditions on an intact and a reduced periodontium: Consensus report of workgroup 1 of the 2017 World Workshop on the Classification of Periodontal and Peri-Implant Diseases and Conditions. J. Periodontol. 2018, 89 (Suppl. S1), S74–S84. [Google Scholar] [CrossRef] [PubMed]
  71. Loe, H.; Theilade, E.; Jensen, S.B. Experimental Gingivitis in Man. J. Periodontol. 1965, 36, 177–187. [Google Scholar] [CrossRef]
  72. Löe, H.; Anerud, A.; Boysen, H.; Morrison, E. Natural history of periodontal disease in man. Rapid, moderate and no loss of attachment in Sri Lankan laborers 14 to 46 years of age. J. Clin. Periodontol. 1986, 13, 431–445. [Google Scholar] [CrossRef] [PubMed]
  73. Lundtorp Olsen, C.; Massarenti, L.; Vendius, V.F.D.; Gürsoy, U.K.; Van Splunter, A.; Bikker, F.J.; Gürsoy, M.; Damgaard, C.; Markvart, M.; Belstrøm, D. Probiotics Support Resilience of the Oral Microbiota during Resolution after Experimental Gingivitis-A Randomized, Double-Blinded, Placebo-Controlled Trial. Nutrients 2023, 15, 4805. [Google Scholar] [CrossRef] [PubMed]
  74. Keller, M.K.; Brandsborg, E.; Holmstrøm, K.; Twetman, S. Effect of tablets containing probiotic candidate strains on gingival inflammation and composition of the salivary microbiome: A randomised controlled trial. Benef. Microbes 2018, 9, 487–494. [Google Scholar] [CrossRef] [PubMed]
  75. Montero, E.; Iniesta, M.; Rodrigo, M.; Marín, M.J.; Figuero, E.; Herrera, D.; Sanz, M. Clinical and microbiological effects of the adjunctive use of probiotics in the treatment of gingivitis: A randomized controlled clinical trial. J. Clin. Periodontol. 2017, 44, 708–716. [Google Scholar] [CrossRef] [PubMed]
  76. Hallström, H.; Lindgren, S.; Yucel-Lindberg, T.; Dahlén, G.; Renvert, S.; Twetman, S. Effect of probiotic lozenges on inflammatory reactions and oral biofilm during experimental gingivitis. Acta Odontol. Scand. 2013, 71, 828–833. [Google Scholar] [CrossRef] [PubMed]
  77. Iniesta, M.; Herrera, D.; Montero, E.; Zurbriggen, M.; Matos, A.R.; Marín, M.J.; Sánchez-Beltrán, M.C.; Llama-Palacio, A.; Sanz, M. Probiotic effects of orally administered Lactobacillus reuteri-containing tablets on the subgingival and salivary microbiota in patients with gingivitis. A randomized clinical trial. J. Clin. Periodontol. 2012, 39, 736–744. [Google Scholar] [CrossRef] [PubMed]
  78. Nowicki, E.M.; Shroff, R.; Singleton, J.A.; Renaud, D.E.; Wallace, D.; Drury, J.; Zirnheld, J.; Colleti, B.; Ellington, A.D.; Lamont, R.J.; et al. Microbiota and Metatranscriptome Changes Accompanying the Onset of Gingivitis. mBio 2018, 9, e00575-18. [Google Scholar] [CrossRef]
  79. Bamashmous, S.; Kotsakis, G.A.; Kerns, K.A.; Leroux, B.G.; Zenobia, C.; Chen, D.; Trivedi, H.M.; McLean, J.S.; Darveau, R.P. Human variation in gingival inflammation. Proc. Natl. Acad. Sci. USA 2021, 118, e2012578118. [Google Scholar] [CrossRef] [PubMed]
  80. Nascimento, G.G.; Danielsen, B.; Baelum, V.; Lopez, R. Identification of inflammatory response patterns in experimental gingivitis studies. Eur. J. Oral Sci. 2019, 127, 33–39. [Google Scholar] [CrossRef] [PubMed]
  81. Escapa, I.F.; Chen, T.; Huang, Y.; Gajare, P.; Dewhirst, F.E.; Lemon, K.P. New Insights into Human Nostril Microbiome from the Expanded Human Oral Microbiome Database (eHOMD): A Resource for the Microbiome of the Human Aerodigestive Tract. mSystems 2018, 3, e00187-18. [Google Scholar] [CrossRef] [PubMed]
  82. Belstrøm, D.; Holmstrup, P.; Bardow, A.; Kokaras, A.; Fiehn, N.E.; Paster, B.J. Temporal Stability of the Salivary Microbiota in Oral Health. PLoS ONE 2016, 11, e0147472. [Google Scholar] [CrossRef] [PubMed]
  83. Hall, M.W.; Singh, N.; Ng, K.F.; Lam, D.K.; Goldberg, M.B.; Tenenbaum, H.C.; Neufeld, J.D.; GBeiko, R.; Senadheera, D.B. Inter-personal diversity and temporal dynamics of dental, tongue, and salivary microbiota in the healthy oral cavity. NPJ Biofilms Microbiomes 2017, 3, 2. [Google Scholar] [CrossRef] [PubMed]
  84. Wirth, R.; Maróti, G.; Lipták, L.; Mester, M.; Al Ayoubi, A.; Pap, B.; Madléna, M.; Minárovits, J.; Kovács, K.L. Microbiomes in supragingival biofilms and saliva of adolescents with gingivitis and gingival health. Oral Dis. 2022, 28, 2000–2014. [Google Scholar] [CrossRef] [PubMed]
  85. Lundtorp Olsen, C.; Markvart, M.; Vendius, V.F.D.; Damgaard, C.; Belstrøm, D. Short-term sugar stress induces compositional changes and loss of diversity of the supragingival microbiota. J. Oral Microbiol. 2023, 15, 2189770. [Google Scholar] [CrossRef] [PubMed]
  86. Yuan, X.; Zhou, F.; Wang, H.; Xu, X.; Xu, S.; Zhang, C.; Zhang, Y.; Lu, M.; Zhang, Y.; Zhou, M.; et al. Systemic antibiotics increase microbiota pathogenicity and oral bone loss. Int. J. Oral Sci. 2023, 15, 4. [Google Scholar] [CrossRef] [PubMed]
  87. Lundtorp-Olsen, C.; Enevold, C.; Twetman, S.; Belstrøm, D. Probiotics Do Not Alter the Long-Term Stability of the Supragingival Microbiota in Healthy Subjects: A Randomized Controlled Trial. Pathogens 2021, 10, 391. [Google Scholar] [CrossRef] [PubMed]
  88. Lundtorp-Olsen, C.; Enevold, C.; Juel Jensen, C.A.; Stofberg, S.N.; Twetman, S.; Belstrøm, D. Impact of Probiotics on the Salivary Microbiota and Salivary Levels of Inflammation-Related Proteins during Short-Term Sugar Stress: A Randomized Controlled Trial. Pathogens 2021, 10, 392. [Google Scholar] [CrossRef] [PubMed]
  89. Ferrer, M.D.; López-López, A.; Nicolescu, T.; Perez-Vilaplana, S.; Boix-Amorós, A.; Dzidic, M.; Garcia, S.; Artacho, A.; Llena, C.; Mira, A. Topic Application of the Probiotic Streptococcus dentisani Improves Clinical and Microbiological Parameters Associated with Oral Health. Front Cell Infect. Microbiol. 2020, 10, 465. [Google Scholar] [CrossRef] [PubMed]
  90. Alanzi, A.; Honkala, S.; Honkala, E.; Varghese, A.; Tolvanen, M.; Söderling, E. Effect of Lactobacillus rhamnosus and Bifidobacterium lactis on gingival health, dental plaque, and periodontopathogens in adolescents: A randomised placebo-controlled clinical trial. Benef. Microbes 2018, 9, 593–602. [Google Scholar] [CrossRef] [PubMed]
  91. Tobita, K.; Watanabe, I.; Tomokiyo, M.; Saito, M. Effects of heat-treated Lactobacillus crispatus KT-11 strain consumption on improvement of oral cavity environment: A randomised double-blind clinical trial. Benef. Microbes 2018, 9, 585–592. [Google Scholar] [CrossRef]
  92. Toiviainen, A.; Jalasvuori, H.; Lahti, E.; Gursoy, U.; Salminen, S.; Fontana, M.; Flannagan, S.; Eckert, G.; Kokaras, A.; Paster, B.; et al. Impact of orally administered lozenges with Lactobacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis BB-12 on the number of salivary mutans streptococci, amount of plaque, gingival inflammation and the oral microbiome in healthy adults. Clin. Oral Investig. 2015, 19, 77–83. [Google Scholar] [CrossRef] [PubMed]
  93. Sinkiewicz, G.; Cronholm, S.; Ljunggren, L.; Dahlén, G.; Bratthall, G. Influence of dietary supplementation with Lactobacillus reuteri on the oral flora of healthy subjects. Swed. Dent. J. 2010, 34, 197–206. [Google Scholar] [PubMed]
  94. Mayanagi, G.; Kimura, M.; Nakaya, S.; Hirata, H.; Sakamoto, M.; Benno, Y.; Shimauchi, H. Probiotic effects of orally administered Lactobacillus salivarius WB21-containing tablets on periodontopathic bacteria: A double-blinded, placebo-controlled, randomized clinical trial. J. Clin. Periodontol. 2009, 36, 506–513. [Google Scholar] [CrossRef]
  95. Könönen, E.; Paju, S.; Pussinen, P.J.; Hyvönen, M.; Di Tella, P.; Suominen-Taipale, L.; Knuuttila, M. Population-based study of salivary carriage of periodontal pathogens in adults. J. Clin. Microbiol. 2007, 45, 2446–2451. [Google Scholar] [CrossRef]
  96. Damgaard, C.; Danielsen, A.K.; Enevold, C.; Massarenti, L.; Nielsen, C.H.; Holmstrup, P.; Belstrøm, D. Porphyromonas gingivalis in saliva associates with chronic and aggressive periodontitis. J. Oral Microbiol. 2019, 11, 1653123. [Google Scholar] [CrossRef] [PubMed]
  97. Tanaka, S.; Murakami, Y.; Seto, K.; Takamori, K.; Yosida, M.; Ochiai, K.; Watanabe, S.; Fujisawa, S. The detection of Porphyromonas gingivalis, Prevotella intermedia, and Actinobacillus actinomycetemcomitans in the supragingival plaque of children with and without caries. Pediatr. Dent. 2003, 25, 143–148. [Google Scholar]
  98. Tanaka, S.; Murakami, Y.; Ogiwara, T.; Shoji, M.; Seto, K.; Nagasaki, M.; Fujisawa, S. Frequency of reactivity for Porphyromonas gingivalis and Prevotella spp. in supra- and subgingival plaques, and periodontal clinical parameters according to subject age. J. Periodontol. 2002, 73, 877–885. [Google Scholar] [CrossRef] [PubMed]
  99. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10 (Suppl. S1), S49–S66. [Google Scholar] [CrossRef] [PubMed]
  100. Bermudez-Brito, M.; Plaza-Díaz, J.; Muñoz-Quezada, S.; Gómez-Llorente, C.; Gil, A. Probiotic mechanisms of action. Ann. Nutr. Metab. 2012, 61, 160–174. [Google Scholar] [CrossRef] [PubMed]
  101. Adams, S.E.; Arnold, D.; Murphy, B.; Carroll, P.; Green, A.K.; Smith, A.M.; Marsh, P.D.; Chen, T.; Marriott, R.E.; Brading, M.G. A randomised clinical study to determine the effect of a toothpaste containing enzymes and proteins on plaque oral microbiome ecology. Sci. Rep. 2017, 7, 43344. [Google Scholar] [CrossRef]
  102. Carda-Diéguez, M.; Moazzez, R.; Mira, A. Functional changes in the oral microbiome after use of fluoride and arginine containing dentifrices: A metagenomic and metatranscriptomic study. Microbiome 2022, 10, 159. [Google Scholar] [CrossRef]
  103. Papadimitriou, K.; Zoumpopoulou, G.; Foligné, B.; Alexandraki, V.; Kazou, M.; Pot, B.; Tsakalidou, E. Discovering probiotic microorganisms: In vitro, in vivo, genetic and omics approaches. Front. Microbiol. 2015, 6, 58. [Google Scholar] [CrossRef] [PubMed]
  104. Davis, C. Enumeration of probiotic strains: Review of culture-dependent and alternative techniques to quantify viable bacteria. J. Microbiol. Methods 2014, 103, 9–17. [Google Scholar] [CrossRef] [PubMed]
  105. Zaura, E.; Brandt, B.W.; Prodan, A.; Teixeira de Mattos, M.J.; Imangaliyev, S.; Kool, J.; Buijs, M.J.; Jagers, F.L.; Hennequin-Hoenderdos, N.L.; Slot, D.E.; et al. On the ecosystemic network of saliva in healthy young adults. ISME J. 2017, 11, 1218–1231. [Google Scholar] [CrossRef] [PubMed]
  106. Segata, N.; Haake, S.K.; Mannon, P.; Lemon, K.P.; Waldron, L.; Gevers, D.; Huttenhower, C.; Izard, J. Composition of the adult digestive tract bacterial microbiome based on seven mouth surfaces, tonsils, throat and stool samples. Genome Biol. 2012, 13, R42. [Google Scholar] [CrossRef]
  107. Kazarina, A.; Kuzmicka, J.; Bortkevica, S.; Zayakin, P.; Kimsis, J.; Igumnova, V.; Sadovska, D.; Freimane, L.; Kivrane, A.; Namina, A.; et al. Oral microbiome variations related to ageing: Possible implications beyond oral health. Arch. Microbiol. 2023, 205, 116. [Google Scholar] [CrossRef] [PubMed]
  108. Kumar, P.S. Oral microbiota and systemic disease. Anaerobe 2013, 24, 90–93. [Google Scholar] [CrossRef]
  109. Peng, X.; Cheng, L.; You, Y.; Tang, C.; Ren, B.; Li, Y.; Xu, X.; Zhou, X. Oral microbiota in human systematic diseases. Int. J. Oral Sci. 2022, 14, 14. [Google Scholar] [CrossRef]
  110. Nyvad, B.; Takahashi, N. Integrated hypothesis of dental caries and periodontal diseases. J. Oral Microbiol. 2020, 12, 1710953. [Google Scholar] [CrossRef] [PubMed]
  111. Nazir, M.; Al-Ansari, A.; Al-Khalifa, K.; Alhareky, M.; Gaffar, B.; Almas, K. Global Prevalence of Periodontal Disease and Lack of Its Surveillance. Sci. World J. 2020, 2020, 2146160. [Google Scholar] [CrossRef] [PubMed]
  112. Mateo, J.; Steuten, L.; Aftimos, P.; André, F.; Davies, M.; Garralda, E.; Geissler, J.; Husereau, D.; Martinez-Lopez, I.; Normanno, N.; et al. Delivering precision oncology to patients with cancer. Nat. Med. 2022, 28, 658–665. [Google Scholar] [CrossRef] [PubMed]
  113. Rosier, B.T.; De Jager, M.; Zaura, E.; Krom, B.P. Historical and contemporary hypotheses on the development of oral diseases: Are we there yet? Front. Cell Infect. Microbiol. 2014, 4, 92. [Google Scholar] [CrossRef] [PubMed]
  114. Socransky, S.S.; Haffajee, A.D.; Cugini, M.A.; Smith, C.; Kent, R.L., Jr. Microbial complexes in subgingival plaque. J. Clin. Periodontol. 1998, 25, 134–144. [Google Scholar] [CrossRef] [PubMed]
  115. Hajishengallis, G.; Darveau, R.P.; Curtis, M.A. The keystone-pathogen hypothesis. Nat. Rev. Microbiol. 2012, 10, 717–725. [Google Scholar] [CrossRef]
  116. Bostanci, N.; Belibasakis, G.N. Porphyromonas gingivalis: An invasive and evasive opportunistic oral pathogen. FEMS Microbiol. Lett. 2012, 333, 1–9. [Google Scholar] [CrossRef] [PubMed]
  117. Hajishengallis, G.; Liang, S.; Payne, M.A.; Hashim, A.; Jotwani, R.; Eskan, M.A.; McIntosh, M.L.; Alsam, A.; Kirkwood, K.L.; Lambris, J.D.; et al. Low-abundance biofilm species orchestrates inflammatory periodontal disease through the commensal microbiota and complement. Cell Host. Microbe 2011, 10, 497–506. [Google Scholar] [CrossRef] [PubMed]
  118. Duran-Pinedo, A.E.; Baker, V.D.; Frias-Lopez, J. The periodontal pathogen Porphyromonas gingivalis induces expression of transposases and cell death of Streptococcus mitis in a biofilm model. Infect. Immun. 2014, 82, 3374–3382. [Google Scholar] [CrossRef] [PubMed]
  119. Duran-Pinedo, A.E.; Solbiati, J.; Teles, F.; Frias-Lopez, J. Subgingival host-microbiome metatranscriptomic changes following scaling and root planing in grade II/III periodontitis. J. Clin. Periodontol. 2023, 50, 316–330. [Google Scholar] [CrossRef]
  120. Luo, S.C.; Wei, S.M.; Luo, X.T.; Yang, Q.Q.; Wong, K.H.; Cheung, P.C.K.; Zhang, B.B. How probiotics, prebiotics, synbiotics, and postbiotics prevent dental caries: An oral microbiota perspective. NPJ Biofilms Microbiomes 2024, 10, 14. [Google Scholar] [CrossRef] [PubMed]
  121. Mayumi, S.; Kuboniwa, M.; Sakanaka, A.; Hashino, E.; Ishikawa, A.; Ijima, Y.; Amano, A. Potential of Prebiotic D-Tagatose for Prevention of Oral Disease. Front. Cell Infect. Microbiol. 2021, 11, 767944. [Google Scholar] [CrossRef]
  122. Tailor, R.; Medara, N.; Chopra, A.; Swarnamali, H.; Eberhard, J.; Jayasinghe, T.N. Role of prebiotic dietary fiber in periodontal disease: A systematic review of animal studies. Front. Nutr. 2023, 10, 1130153. [Google Scholar] [CrossRef] [PubMed]
  123. Cuschieri, S. The STROBE guidelines. Saudi J. Anaesth. 2019, 13 (Suppl. S1), S31–S34. [Google Scholar] [CrossRef] [PubMed]
  124. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Oral probiotics aim to interact with the host microbiome to support oral health and halt the progression of oral diseases.
Figure 1. Oral probiotics aim to interact with the host microbiome to support oral health and halt the progression of oral diseases.
Pathogens 13 00419 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lundtorp-Olsen, C.; Markvart, M.; Twetman, S.; Belstrøm, D. Effect of Probiotic Supplements on the Oral Microbiota—A Narrative Review. Pathogens 2024, 13, 419. https://doi.org/10.3390/pathogens13050419

AMA Style

Lundtorp-Olsen C, Markvart M, Twetman S, Belstrøm D. Effect of Probiotic Supplements on the Oral Microbiota—A Narrative Review. Pathogens. 2024; 13(5):419. https://doi.org/10.3390/pathogens13050419

Chicago/Turabian Style

Lundtorp-Olsen, Christine, Merete Markvart, Svante Twetman, and Daniel Belstrøm. 2024. "Effect of Probiotic Supplements on the Oral Microbiota—A Narrative Review" Pathogens 13, no. 5: 419. https://doi.org/10.3390/pathogens13050419

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop